Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Clin Chim Acta ; 398(1-2): 48-52, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18727923

RESUMEN

BACKGROUND: The discovery of molecular biomarkers is crucial to the diagnosis of head and neck squamous cell cancer (HNSCC). METHODS: Proteins from pre-surgery serum samples of patients with HNSCC and healthy individuals were analyzed by 2-dimensional gel electrophoresis (2-DE) using a 17 cm-long immobilized pH gradient gel strip (large gel). The differentially expressed protein spots were detected by statistical analysis. Because 2 haptoglobin (Hp) alpha chains were found to be differentially expressed, the genotypic distribution of Hp alpha chains in patients and healthy individuals was assayed by polymerase chain reaction. The protein expression levels of Hp alpha chains in individuals carrying different Hp alleles were analyzed by 2-DE with a small gel. RESULTS: Two isoforms of haptoglobin alpha2 chain (Hp alpha2) in patients' sera were found from 2-DE analysis to be up-regulated, while the isoforms of haptoglobin alpha1 chain (Hp alpha1) were significantly down-regulated. Apolipoprotein AII and 2 isoforms of apolipoprotein CII were also differentially expressed in the sera of patients with HNSCC. The Hp alpha2 chain was significantly up-regulated in the patients carrying at least one haptoglobin 2 allele, according to the spot intensities from scanned images of small-gel 2-DE. CONCLUSIONS: The expression pattern of seven differentially expressed polypeptides and the up-regulation of Hp alpha2 in individuals with the Hp 2 allele are potential biomarkers.


Asunto(s)
Haptoglobinas/metabolismo , Neoplasias de Cabeza y Cuello/diagnóstico , Neoplasias de Células Escamosas/diagnóstico , Adolescente , Anciano , Alelos , Apolipoproteína A-II/biosíntesis , Apolipoproteína A-II/genética , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Biomarcadores , Electroforesis en Gel Bidimensional , Electroforesis en Gel de Poliacrilamida , Femenino , Genómica , Haptoglobinas/análisis , Haptoglobinas/genética , Neoplasias de Cabeza y Cuello/sangre , Neoplasias de Cabeza y Cuello/genética , Humanos , Concentración de Iones de Hidrógeno , Focalización Isoeléctrica , Isomerismo , Masculino , Persona de Mediana Edad , Neoplasias de Células Escamosas/sangre , Neoplasias de Células Escamosas/genética , Proteoma/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tinción con Nitrato de Plata , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Adulto Joven
2.
Biochem J ; 397(1): 159-68, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16584384

RESUMEN

In the present study, we have characterized signalling cross-talk between STAT5b (signal transducer and activator of transcription 5b) and HNF4alpha (hepatocyte nuclear factor 4alpha), two major regulators of sex-dependent gene expression in the liver. In a HepG2 liver cell model, HNF4alpha strongly inhibited beta-casein and ntcp (Na+/taurocholate cotransporting polypeptide) promoter activity stimulated by GH (growth hormone)-activated STAT5b, but had no effect on interferon-gamma-stimulated STAT1 transcriptional activity. By contrast, STAT5b synergistically enhanced the transcriptional activity of HNF4alpha towards the ApoCIII (apolipoprotein CIII) promoter. The inhibitory effect of HNF4alpha on STAT5b transcription was associated with the inhibition of GH-stimulated STAT5b tyrosine phosphorylation and nuclear translocation. The short-chain fatty acid, butyrate, reversed STAT5b transcriptional inhibition by HNF4alpha, but did not reverse the inhibition of STAT5b tyrosine phosphorylation. HNF4alpha inhibition of STAT5b tyrosine phosphorylation was not reversed by pervanadate or by dominant-negative phosphotyrosine phosphatase 1B, suggesting that it does not result from an increase in STAT5b dephosphorylation. Rather, HNF4alpha blocked GH-stimulated tyrosine phosphorylation of JAK2 (Janus kinase 2), a STAT5b tyrosine kinase. Thus STAT5b and HNF4alpha exhibit bi-directional cross-talk that may augment HNF4alpha-dependent gene transcription while inhibiting STAT5b transcriptional activity via the inhibitory effects of HNF4alpha on JAK2 phosphorylation, which leads to inhibition of STAT5b signalling initiated by the GH receptor at the cell surface.


Asunto(s)
Factor Nuclear 4 del Hepatocito/fisiología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Somatotropina/fisiología , Factor de Transcripción STAT5/fisiología , Animales , Apolipoproteína C-III , Apolipoproteínas C/biosíntesis , Células COS , Carcinoma Hepatocelular , Caseínas , Chlorocebus aethiops , Femenino , Factor Nuclear 4 del Hepatocito/genética , Humanos , Janus Quinasa 2 , Hígado/enzimología , Neoplasias Hepáticas , Masculino , Ratones , Fosforilación , Regiones Promotoras Genéticas , Ratas , Receptor Cross-Talk , Factor de Transcripción STAT5/genética , Factores Sexuales , Transcripción Genética , Activación Transcripcional , Células Tumorales Cultivadas
3.
J Clin Invest ; 115(5): 1290-7, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15841205

RESUMEN

CD36 mediates the transfer of fatty acids (FAs) across the plasma membranes of muscle and adipose cells, thus playing an important role in regulating peripheral FA metabolism in vivo. In the proximal intestine, CD36 is localized in abundant quantities on the apical surface of epithelial cells, a pattern similar to that of other proteins implicated in the uptake of dietary FAs. To define the role of CD36 in the intestine, we examined FA utilization and lipoprotein secretion by WT and CD36-null mice in response to acute and chronic fat feeding. CD36-null mice given a fat bolus by gavage or fed a high-fat diet accumulated neutral lipid in the proximal intestine, which indicated abnormal lipid processing. Using a model in which mice were equipped with lymph fistulae, we obtained evidence of defective lipoprotein secretion by directly measuring lipid output. The secretion defect appeared to reflect an impaired ability of CD36-null enterocytes to efficiently synthesize triacylglycerols from dietary FAs in the endoplasmic reticulum. In the plasma of intact mice, the reduced intestinal lipid secretion was masked by slow clearance of intestine-derived lipoproteins. The impaired clearance occurred despite normal lipoprotein lipase activity and likely reflected feedback inhibition of the lipase by FAs due to their defective removal from the plasma. We conclude that CD36 is important for both secretion and clearance of intestinal lipoproteins. CD36 deficiency results in hypertriglyceridemia both in the postprandial and fasting states and in humans may constitute a risk factor for diet-induced type 2 diabetes and cardiovascular disease.


Asunto(s)
Antígenos CD36/metabolismo , Quilomicrones/sangre , Intestino Delgado/metabolismo , Metabolismo de los Lípidos , Animales , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Antígenos CD36/genética , Enterocitos/metabolismo , Ácidos Grasos/metabolismo , Intestino Delgado/patología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Factores de Tiempo , Triglicéridos/metabolismo
4.
Atherosclerosis ; 178(2): 257-64, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15694932

RESUMEN

ApoC-I plays an important role in controlling plasma lipid metabolism, however little is known about factors regulating the hepatic synthesis and secretion of this apolipoprotein. In the present study, we have carried out experiments with human hepatoma (HepG2) cells, in order to determine the effect of different tissue culture conditions on cellular lipid levels and on the production of apoC-I (and apoE) at the protein and mRNA level. Cells incubated for 48 h with 10% human serum had significantly higher cellular triglyceride (22%, P<0.05) and cholesterol levels (19%, P<0.01), higher medium apoC-I and apoE levels (2.6- and 2.9-fold, respectively), but similar levels of apoC-I and apoE mRNA, compared to cells incubated with 10% human lipoprotein-deficient serum (LPDS). Serum containing only HDL, or containing HDL with LDL, also increased cellular lipids and increased secreted apoC-I and apoE levels without altering apoC-I and apoE mRNA levels. Incubation of cells with Intralipid triglyceride (625 microM), increased cellular triglyceride (2.8-fold, P<0.001), decreased cellular cholesterol (32%, P<0.01), decreased cellular and medium apoC-I (24 and 26%, P<0.01) and had no effect on apoC-I mRNA levels. Additional experiments in which cells were loaded with cholesterol (incubation with 10 microg/ml cholesterol plus 1 microg/ml 25-hydroxycholesterol) or depleted of cholesterol (statin treatment) confirmed that secretion of apoC-I by HepG2 cells was dependent on cellular cholesterol levels and independent of changes in apoC-I mRNA levels. These results demonstrate that cellular cholesterol rather than triglyceride levels play a role in controlling apoC-I production by HepG2 cells and that this regulation occurs at a post-transcriptional level.


Asunto(s)
Apolipoproteínas C/biosíntesis , Apolipoproteínas C/metabolismo , Arteriosclerosis/fisiopatología , Colesterol/farmacología , Apolipoproteína C-I , Carcinoma Hepatocelular/patología , Humanos , Líquido Intracelular/química , Neoplasias Hepáticas/patología , Procesamiento Postranscripcional del ARN , ARN Mensajero/biosíntesis , Células Tumorales Cultivadas
5.
J Biol Chem ; 280(13): 12559-66, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15649902

RESUMEN

The apoAI/CIII/AIV gene cluster is involved in lipid metabolism and has a complex pattern of gene expression modulated by a common regulatory element, the apoCIII enhancer. A new member of this cluster, apolipoprotein (apo) AV, has recently been discovered as a novel modifier in triglyceride metabolism. To determine the expression of all four apo genes in combination and, most importantly, whether the transcription of apoAV is coregulated by the apoCIII enhancer in the cluster, we generated an intact transgenic line carrying the 116-kb human apoAI/CIII/AIV/AV gene cluster and a mutant transgenic line in which the apoCIII enhancer was deleted from the 116-kb structure. We demonstrated that the apoCIII enhancer regulated hepatic and intestinal apoAI, apoCIII, and apoAIV expression; however, it did not direct the newly identified apoAV in the cluster. Furthermore, human apo genes displayed integrated position-independent expression and a closer approximation of copy number-dependent expression in the intact transgenic mice. Because apoCIII and apoAV play opposite roles in triglyceride homeostasis, we analyzed the lipid profiles in our transgenic mice to assess the effects of human apoAI gene cluster expression on lipid metabolism. The triglyceride level was elevated in intact transgenic mice but decreased in mutant ones compared with nontransgenic mice. In addition, the expression of human apoAI and apoAIV elevated high density lipoprotein cholesterol in transgenic mice fed an atherogenic diet. In conclusion, our studies with human apoAI/CIII/AIV/AV gene cluster transgenic models showed that the apoCIII enhancer regulated expression of apoAI, apo-CIII, and apoAIV but not apoAV in vivo and showed the influences of expression of the entire cluster on lipid metabolism.


Asunto(s)
Apolipoproteína A-I/biosíntesis , Apolipoproteína A-I/genética , Apolipoproteínas A/biosíntesis , Apolipoproteínas A/genética , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Familia de Multigenes , Animales , Apolipoproteína C-III , Arteriosclerosis , Western Blotting , HDL-Colesterol/metabolismo , Cromosomas Artificiales Bacterianos , ADN/metabolismo , Dieta Aterogénica , Eliminación de Gen , Humanos , Hibridación Fluorescente in Situ , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Ratones Transgénicos , Modelos Genéticos , Mutación , ARN/metabolismo , Factores de Tiempo , Distribución Tisular , Transcripción Genética , Triglicéridos/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 288(6): G1150-8, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15662048

RESUMEN

Essential fatty acid (EFA) deficiency in mice decreases plasma triglyceride (TG) concentrations and increases hepatic TG content. We evaluated in vivo and in vitro whether decreased hepatic secretion of TG-rich very low-density lipoprotein (VLDL) contributes to this consequence of EFA deficiency. EFA deficiency was induced in mice by feeding an EFA-deficient (EFAD) diet for 8 wk. Hepatic VLDL secretion was quantified in fasted EFAD and EFA-sufficient (EFAS) mice using the Triton WR-1339 method. In cultured hepatocytes from EFAD and EFAS mice, VLDL secretion into medium was measured by quantifying [(3)H]-labeled glycerol incorporation into TG and phospholipids. Hepatic expression of genes involved in VLDL synthesis and clearance was measured, as were plasma activities of lipolytic enzymes. TG secretion rates were quantitatively similar in EFAD and EFAS mice in vivo and in primary hepatocytes from EFAD and EFAS mice in vitro. However, EFA deficiency increased the size of secreted VLDL particles, as determined by calculation of particle diameter, particle sizing by light scattering, and evaluation of the TG-to-apoB ratio. EFA deficiency did not inhibit hepatic lipase and lipoprotein lipase activities in plasma, but increased hepatic mRNA levels of apoAV and apoCII, both involved in control of lipolytic degradation of TG-rich lipoproteins. EFA deficiency does not affect hepatic TG secretion rate in mice, but increases the size of secreted VLDL particles. Present data suggest that hypotriglyceridemia during EFA deficiency is related to enhanced clearance of altered VLDL particles.


Asunto(s)
Ácidos Grasos Esenciales/deficiencia , Hígado Graso/etiología , Hígado Graso/fisiopatología , Lipoproteínas VLDL/metabolismo , Hígado/fisiología , Animales , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/farmacología , Dieta , Perfilación de la Expresión Génica , Lipasa/farmacología , Masculino , Ratones , Tamaño de la Partícula , ARN Mensajero/biosíntesis , Triglicéridos/sangre
7.
J Nutr ; 134(11): 2935-41, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15514255

RESUMEN

Little is known about the regulation of apolipoprotein (apo) C-I production by human adipocytes. The aim of the present study, therefore, was to investigate the effect of different tissue culture conditions on the synthesis and secretion of apoC-I and apoE in human SW872 liposarcoma cells. After 3-4 d in culture (0.5 x 10(6) cells/well, DMEM/F-12 medium with 10% fetal calf serum), cells reached confluence and became growth arrested. The molar ratio of apoE:apoC-I in the cell was 8.9 +/- 0.6 and in the medium was 6.6 +/- 0.5. After 17 d in culture, SW872 cells contained significantly more cholesterol (100%) and triglyceride (3-fold) and secreted more apoC-I [4 vs. 17 d: 0.11 +/- 0.01 vs. 0.23 +/- 0.01 pmol/(10(6) cells . 24 h), P < 0.001] and apoE [0.7 +/- 0.1 vs. 3.1 +/- 0.3 pmol/(10(6) cells . 24 h), P < 0.001]. Cellular apoC-I increased 7-fold and apoE increased 16-fold. Cell maturation was associated with significantly higher levels of apoE mRNA but not apoC-I mRNA. Increases in cell lipids, apoC-I, and apoE were not dependent on the presence of extracellular lipids because similar changes occurred in cells incubated with lipoprotein-deficient serum or in cells incubated without serum. Treatment (7 d) of cells during maturation with insulin (10 or 1000 nmol/L) significantly reduced the secretion of apoC-I and apoE. These results demonstrate that in maturing SW872 cells, cholesterol and triglyceride accumulation in the presence or absence of extracellular lipids, is associated with increased apoC-I and apoE production. Furthermore, apoC-I and apoE production are differentially regulated at the transcriptional level, and long-term treatment with insulin has an inhibitory rather than stimulatory effect on apoC-I and apoE production.


Asunto(s)
Adipocitos/metabolismo , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/metabolismo , Apolipoproteínas E/biosíntesis , Apolipoproteínas E/metabolismo , Adipocitos/química , Adipocitos/efectos de los fármacos , Apolipoproteínas C/genética , Apolipoproteínas E/genética , División Celular , Colesterol/metabolismo , Medios de Cultivo , Humanos , Insulina/farmacología , Liposarcoma , PPAR alfa/genética , PPAR gamma/genética , ARN Mensajero/análisis , Triglicéridos/metabolismo , Células Tumorales Cultivadas
8.
Atherosclerosis ; 177(1): 137-45, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15488876

RESUMEN

Apolipoprotein (apo) C-III plays an important role in regulating plasma triglyceride (TG) metabolism. In order to further investigate the plasma metabolism of apoC-III in hypertriglyceridemic subjects, we have studied the plasma kinetics of VLDL apoC-III, HDL apoC-III and total plasma apoC-III with a primed constant intravenous infusion of deuterated leucine in a group of male patients with mixed hyperlipidemia (type IIb hyperlipoproteinemia, HLP, n=6) and in a group with type III HLP (n=6). Compared to normolipidemic control subjects (n=5), patients with type IIb and type III HLP had significantly higher levels of plasma TG (0.89 +/- 0.15 mmol/l vs 2.56 +/- 0.40 mmol/l vs 8.76 +/- 1.39 mmol/l, respectively, P <0.01), plasma apoC-III (9.5 +/- 0.8 mg/dl vs 20.8 +/- 2.5 mg/dl vs 41.7 +/- 5.6 mg/dl, P <0.01) and VLDL apoC-III (3.6 +/- 0.8 mg/dl vs 14.6 +/- 2.2 mg/dl vs 35.4 +/- 5.1 mg/dl, P <0.01). VLDL apoC-III production rates were significantly elevated in type IIb and type III patients (1.35 +/- 0.23 mg kg(-1) day(-1) vs 3.53 +/- 0.43 mg kg(-1) day(-1) vs 5.60 +/- 0.78 mg kg(-1) day(-1), P <0.01), as were total plasma apoC-III production rates (1.80 +/- 0.22 mg kg(-1) day(-1) vs 4.16 +/- 0.44 mg kg(-1) day(-1) vs 7.26 +/- 0.74 mg kg(-1) day(-1), P <0.01). VLDL apoC-III but not total plasma apoC-III fractional catabolic rates were reduced in type IIb and type III patients. Together with our previous results showing an increase of apoC-III production in patients with type IV HLP, and in overweight subjects with reduced insulin sensitivity, our data suggest that increased apoC-III production is a characteristic feature of patients with hypertriglyceridemia.


Asunto(s)
Apolipoproteínas C/biosíntesis , Hipertrigliceridemia/metabolismo , Adulto , Apolipoproteína C-III , Apolipoproteínas C/sangre , Humanos , Hipertrigliceridemia/sangre , Lipoproteínas VLDL/sangre , Persona de Mediana Edad
9.
J Lipid Res ; 45(12): 2235-44, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15342689

RESUMEN

The adaptive value of apolipoprotein B-48 (apoB-48), the truncated form of apoB produced by the intestine, in lipid metabolism remains unclear. We crossed human apoC-III transgenic mice with mice expressing either apoB-48 only (apoB48/48) or apoB-100 only (apoB100/100). Cholesterol levels were higher in apoB48/48 mice than in apoB100/100 mice but triglyceride levels were similar. Lipid levels were increased by the apoC-III transgene. However, triglyceride levels were significantly higher in apoB100/100C-III than in apoB48/48C-III mice (895 +/- 395 mg/dl vs. 690 +/- 252 mg/dl; P <0.01), whereas cholesterol levels were higher in the apoB48/48C-III mice than in apoB100/100C-III (144 +/- 35 mg/dl vs. 94 +/- 30 mg/dl; P <0.00001). Triglyceride clearance from VLDL was impaired to a greater extent in apoB100/100C-III vs. apoB100/100 mice than in apoB48/48C-III vs. apoB48/48 mice. Triglyceride secretion rates were no different in apoC-III transgenic mice than in their nontransgenic littermates. ApoB-48 triglyceride-rich lipoproteins were more resistant to the triglyceride-increasing effects of apoC-III but appeared more sensitive to the remnant clearance inhibition. Our findings support a coordinated role for apoB-48 in facilitating the delivery of dietary triglycerides to the periphery. Consistent with such a mechanism, glucose levels were significantly higher in apoB48/48 mice vs. apoB100/100 mice, perhaps on the basis of metabolic competition.


Asunto(s)
Apolipoproteínas B/genética , Apolipoproteínas C/genética , Hipertrigliceridemia/genética , Animales , Apolipoproteína B-100 , Apolipoproteína B-48 , Apolipoproteína C-III , Apolipoproteínas B/metabolismo , Apolipoproteínas C/biosíntesis , VLDL-Colesterol/sangre , Femenino , Marcación de Gen , Hipertrigliceridemia/metabolismo , Masculino , Ratones , Triglicéridos/sangre
10.
J Clin Endocrinol Metab ; 89(8): 3949-55, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15292332

RESUMEN

Overweight individuals with reduced insulin sensitivity often have mild to moderate hypertriglyceridemia. To investigate the role of apolipoprotein (apo)C-III metabolism in the etiology of hypertriglyceridemia in these individuals, we investigated 10 male subjects with different body weights (body mass index, 24-34 kg/m(2)) and insulin sensitivity (homeostasis model assessment, 4.7-35.0). Total plasma and very-low-density lipoprotein (VLDL) apoC-III kinetics, as well as VLDL triglyceride (TG) and VLDL apoB kinetics, were measured with iv injected stable isotopes. The apoC-III, TG, and apoB levels in VLDL ranged from 2.9-18.2 mg/dl, 0.49-2.89 mmol/liter, and 6.7-29.3 mg/dl, respectively. Mean production rates (PRs) were: VLDL apoC-III, 20.2 +/- 4.1 micromol/d (range, 8.0-44.8); VLDL TG, 26.9 +/- 4.6 mmol/d (range, 10.2-51.1); and VLDL apoB, 4.4 +/- 0.8 micromol/d (range, 1.5-9.1). VLDL apoC-III PRs were significantly correlated with body mass index, homeostasis model assessment, and plasma TG (r = 0.66, P < 0.05; r = 0.80, P < 0.01; r = 0.95, P < 0.001, respectively). Similar correlations were found for plasma apoC-III PRs (r = 0.70, P < 0.05; r = 0.67, P < 0.05; r = 0.80, P < 0.01, respectively). Fractional catabolic rates (FCRs) were not significantly related to metabolic variables. VLDL TG levels were strongly related to VLDL apoC-III levels (r = 0.99, P < 0.001) and VLDL apoC-III PRs (r = 0.94, P < 0.001). VLDL apoC-III levels were more strongly correlated with VLDL TG PRs (r = 0.81, P < 0.01) than with VLDL TG FCRs or VLDL apoB FCRs (r = -0.53, P = 0.12; r = -0.37, P = 0.29). These results suggest that increased hepatic production of VLDL apoC-III is characteristic of subjects with higher body weights and lower levels of insulin sensitivity and is strongly related to the plasma concentration and level of production of VLDL TG.


Asunto(s)
Apolipoproteínas C/biosíntesis , Peso Corporal , Resistencia a la Insulina/fisiología , Insulina/fisiología , Lipoproteínas VLDL/sangre , Triglicéridos/sangre , Adulto , Apolipoproteína C-III , Humanos , Cinética , Masculino , Persona de Mediana Edad , Concentración Osmolar
11.
Metabolism ; 53(5): 607-13, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15131765

RESUMEN

Our previous study demonstrated that fenofibrate improves both lipid metabolism and obesity, in part through hepatic peroxisome proliferator-activated receptor alpha (PPARalpha) activation, in female ovariectomized, but not in sham-operated, low-density lipoprotein receptor-null (LDLR-null) mice. The aim of this study was to determine whether fenofibrate prevents obesity and hypertriglyceridemia in male LDLR-null mice. Mice fed a high-fat diet for 8 weeks exhibited increases in body and white adipose tissue (WAT) weights and developed severe hypertriglyceridemia compared with mice fed a low-fat control diet. However, these effects were effectively prevented by fenofibrate. Mice given a fenofibrate-supplemented high-fat diet showed significantly reduced body weight, WAT weight, and serum triglycerides versus high-fat diet-fed animals. Triton WR1339 study showed that fenofibrate-induced reduction in circulating triglycerides was due to the decreased secretion of triglycerides from the liver. Moreover, the administration of fenofibrate not only resulted in liver hypertrophy and reduction in hepatic lipid accumulation, but also regulated the transcriptional expression of PPARalpha target genes, such as hepatic acyl-coenzyme A (CoA) oxidase and apolipoprotein C-III (apoC-III). Therefore, our results suggest that alterations in hepatic PPARalpha action by fenofibrate seem to suppress diet-induced obesity and severe hypertriglyceridemia caused by LDLR deficiency in male mice.


Asunto(s)
Fenofibrato/farmacología , Hipertrigliceridemia/prevención & control , Hipolipemiantes/farmacología , Obesidad/prevención & control , Receptores de LDL/deficiencia , Acil-CoA Oxidasa/biosíntesis , Acil-CoA Oxidasa/genética , Tejido Adiposo/metabolismo , Animales , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Peso Corporal/efectos de los fármacos , Colesterol/sangre , Grasas de la Dieta/metabolismo , Epidídimo/metabolismo , Metabolismo de los Lípidos , Hígado/citología , Hígado/metabolismo , Hígado/ultraestructura , Masculino , Ratones , Ratones Transgénicos , Tamaño de los Órganos/efectos de los fármacos , Polietilenglicoles/farmacología , ARN Mensajero/biosíntesis , Receptores de LDL/genética , Triglicéridos/sangre , Triglicéridos/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 24(4): 637-43, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14751813

RESUMEN

Retinoic acid receptor-related Orphan Receptor alpha (RORalpha) is a member of the nuclear hormone receptor superfamily. RORalpha has long been considered as a constitutive activator of transcription in the absence of exogenous ligand; however, cholesterol has recently been identified as a natural ligand of RORalpha. The spontaneous staggerer (sg/sg) mutation is a deletion in the Rora gene that prevents the translation of the ligand-binding domain (LBD), leading to the loss of RORalpha activity. The homozygous Rora(sg/sg) mutant mouse, of which the most obvious phenotype is ataxia associated with cerebellar degeneration, also displays a variety of other phenotypes, including several vascular ones; in particular, dysfunction of smooth muscle cells and enhanced susceptibility to atherosclerosis. Moreover, RORalpha appears to participate in the regulation of plasma cholesterol levels, and has been shown to positively regulate apolipoprotein (apo)A-I and apoC-III gene expression. Yet its activity is regulated by cholesterol itself, making RORalpha an intracellular cholesterol target.


Asunto(s)
Colesterol/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transactivadores/metabolismo , Animales , Apolipoproteína A-I/biosíntesis , Apolipoproteína A-I/genética , Apolipoproteína C-III , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Arteriosclerosis/genética , Arteriosclerosis/metabolismo , Circulación Colateral/fisiología , Retroalimentación Fisiológica , Regulación de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Humanos , Inflamación/metabolismo , Ratones , Ratones Mutantes Neurológicos , Modelos Biológicos , Músculo Esquelético/metabolismo , Músculo Liso Vascular/fisiopatología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Células de Purkinje/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/metabolismo , Transactivadores/deficiencia , Transactivadores/genética , Transcripción Genética/fisiología
13.
J Biol Chem ; 278(47): 46919-26, 2003 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-12954636

RESUMEN

Apolipoprotein E (apoE) is synthesized in many tissues, and the liver is the primary site from which apoE redistributes cholesterol and other lipids to peripheral tissues. Here we demonstrate that the TR4 orphan nuclear receptor (TR4) can induce apoE expression in HepG2 cells. This TR4-mediated regulation of apoE gene expression was further confirmed in vivo using TR4 knockout mice. Both serum apoE protein and liver apoE mRNA levels were significantly reduced in TR4 knockout mice. Gel shift and luciferase reporter gene assays further demonstrated that TR4 can induce apoE gene expression via a TR4 response element located in the hepatic control region that is 15 kb downstream of the apoE gene. Furthermore our in vivo data from TR4 knockout mice prove that TR4 can also regulate apolipoprotein C-I and C-II gene expression via the TR4 response element within the hepatic control region. Together our data show that loss of TR4 down-regulates expression of the apoE/C-I/C-II gene cluster in liver cells, demonstrating important roles of TR4 in the modulation of lipoprotein metabolism.


Asunto(s)
Apolipoproteínas/biosíntesis , Regulación de la Expresión Génica , Hígado/metabolismo , Familia de Multigenes , Receptores de Esteroides/fisiología , Receptores de Hormona Tiroidea/fisiología , Animales , Apolipoproteína C-I , Apolipoproteína C-II , Apolipoproteínas/genética , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Apolipoproteínas E/biosíntesis , Apolipoproteínas E/genética , Línea Celular Tumoral , Humanos , Hígado/citología , Ratones , Ratones Noqueados , ARN Mensajero/análisis , Receptores de Esteroides/deficiencia , Receptores de Hormona Tiroidea/deficiencia , Elementos de Respuesta , Transfección
14.
J Cardiovasc Pharmacol ; 42(2): 251-7, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12883330

RESUMEN

Apolipoprotein (apo) E and C-I are plasma apolipoproteins that have been implicated in the etiology of atherosclerosis and obesity, respectively. Both proteins are synthesized and secreted by macrophages, though pharmacological regulation of their production is poorly understood. The authors compared the effect of 2 HMG-CoA reductase inhibitors, atorvastatin and cerivastatin, on the synthesis and secretion of apoE and apoC-I by THP-1 macrophages. Atorvastatin reduced medium apoE and cellular apoE mRNA of PMA-activated THP-1 cells in a dose-dependent manner (-24% and -22%, respectively, at 1-micromol/L, P < 0.01). ApoC-I in the medium was also reduced by atorvastatin in a dose-dependent manner, though to a lesser extent (-15% at 1-micromol/L, P < 0.05). Cerivastatin similarly reduced medium apoE (-20% at 1-micromol/L, P < 0.05) and cellular apoE mRNA (-31% at 1-micromol/L, P < 0.05), and significantly lowered cellular apoC-I mRNA (-15%, P < 0.05), but not apoC-I in the medium. In experiments with THP-1 macrophages loaded with cholesterol (ie, 24-hour incubation with acetyl-LDL), atorvastatin and cerivastatin (1-micromol/L) significantly (P < 0.05) reduced both medium apoE (-30% and -25%, respectively) and cellular apoE mRNA (-25% and -17%, respectively). A lower and less consistent effect was observed on medium apoC-I (-6% and -18%, respectively) and cellular apoC-I mRNA (-13% and -19%, respectively). These data demonstrate that statins have the capacity to reduce the synthesis and secretion of both apoE and apoC-I in THP-1 macrophages loaded or unloaded with cholesterol.


Asunto(s)
Apolipoproteínas C/biosíntesis , Apolipoproteínas E/biosíntesis , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Macrófagos/efectos de los fármacos , Pirroles/farmacología , Apolipoproteína C-I , Apolipoproteínas C/metabolismo , Apolipoproteínas E/metabolismo , Atorvastatina , Células Cultivadas , Macrófagos/metabolismo
15.
J Lipid Res ; 43(12): 2136-45, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12454276

RESUMEN

Apolipoprotein C-I (apoC-I) has been proposed to act primarily via interference with apoE-mediated lipoprotein uptake. To define actions of apoC-I that are independent of apoE, we crossed a moderately overexpressing human apoC-I transgenic, which possesses a minimal phenotype in the WT background, with the apoE-null mouse. Surprisingly, apoE-null/C-I mice showed much more severe hyperlipidemia than apoE-null littermates in both the fasting and non-fasting states, with an almost doubling of cholesterol, primarily in IDL+LDL, and a marked increase in triglycerides; 3-fold in females to 260 +/- 80 mg/dl and 14-fold in males to 1409 +/- 594 mg/dl. HDL lipids were not significantly altered but HDL were apoC-I-enriched and apoA-II-depleted. Production rates of VLDL triglyceride were unchanged as was the clearance of post-lipolysis remnant particles. Plasma post-heparin hepatic lipase and lipoprotein lipase levels were undiminished as was the in vitro hydrolysis of apoC-I transgenic VLDL. However, HDL from apoC-I transgenic mice had a marked inhibitory effect on hepatic lipase activity, as did purified apoC-I. LPL activity was minimally affected. Atherosclerosis assay revealed significantly increased atherosclerosis in apoE-null/C-I mice assessed via the en face assay. Inhibition of hepatic lipase may be an important mechanism of the decrease in lipoprotein clearance mediated by apoC-I.


Asunto(s)
Apolipoproteínas C/genética , Apolipoproteínas E/genética , Hipertrigliceridemia/genética , Lipasa/antagonistas & inhibidores , Hígado/metabolismo , Animales , Apolipoproteína C-I , Apolipoproteínas C/biosíntesis , Arteriosclerosis , VLDL-Colesterol/sangre , VLDL-Colesterol/metabolismo , Humanos , Hipertrigliceridemia/enzimología , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
16.
J Lipid Res ; 43(12): 2172-9, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12454280

RESUMEN

Elevated serum levels of triglyceride-rich remnant lipoproteins (TRL) are a major risk factor predisposing a subject to atherosclerosis. Apolipoprotein C-III (apoC-III) is a major constituent of TRL that impedes triglyceride hydrolysis and remnant clearance and, as such, may exert pro-atherogenic activities. In the present study, transient cotransfection experiments in rat hepatocytes in primary culture and rabbit kidney RK13 cells demonstrated that overexpression of Rev-erbalpha specifically decreases basal and HNF-4 stimulated human apoC-III promoter activity. A Rev-erbalpha response element was mapped by promoter deletion, mutation analysis, and gel-shift experiments to a AGGTCA half-site located at position -23/-18 (downstream of the TATA box) in the apoC-III promoter. Finally, Rev-erbalpha-deficient mice displayed elevated serum and liver mRNA levels of apoC-III together with increased serum VLDL triglycerides. Taken together, our data identify Rev-erbalpha as a regulator of apoC-III gene expression, providing a novel, physiological role for this nuclear receptor in the regulation of lipid metabolism.


Asunto(s)
Apolipoproteínas C/metabolismo , Proteínas de Unión al ADN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transcripción Genética/fisiología , Animales , Apolipoproteína C-III , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , VLDL-Colesterol/sangre , VLDL-Colesterol/metabolismo , Regulación de la Expresión Génica/fisiología , Hepatocitos , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos BALB C , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas
17.
Biochemistry ; 41(48): 14313-22, 2002 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-12450397

RESUMEN

We have investigated the effect of disulfide cross-linking on amyloid formation by human apolipoprotein (apo) C-II. Three derivatives of apoC-II were generated by inserting a cysteine residue on either the N-terminus (C(N)-apoC-II), C-terminus (C(C)-apoC-II), or both termini (C(N)C(C)-apoC-II). Under reducing conditions, all derivatives formed amyloid with a fibrous ribbon morphology similar to that of wild-type apoC-II. Under oxidizing conditions, C(N)- and C(N)C(C)-apoC-II formed a highly tangled network of fibrils, suggesting that the addition of an N-terminal cysteine to apoC-II promotes interfibril disulfide cross-links. Fibrils formed by C(C)-apoC-II under oxidizing conditions were closely packed but less tangled than fibrils formed by the C(N) and C(N)C(C) derivatives. The frequency of closed ring structures was more than doubled for C(C)-apoC-II compared to wild-type apoC-II. The kinetics of fibril formation by all cysteine derivatives was markedly enhanced under oxidizing conditions, suggesting that disulfide cross-linking promotes amyloid formation. Substoichiometric levels of preformed C(N)- and C(C)-apoC-II dimers accelerate amyloid formation by wild-type apoC-II. These data suggest that the N- and C-termini of apoC-II are close together in the amyloid fibril such that covalent cross-linking of either the N or C end of apoC-II promotes nucleation and the "seeding" of fibril growth.


Asunto(s)
Amiloide/química , Apolipoproteínas C/química , Reactivos de Enlaces Cruzados/química , Cisteína/química , Fragmentos de Péptidos/química , Amiloide/genética , Amiloide/metabolismo , Amiloide/ultraestructura , Apolipoproteína C-II , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Apolipoproteínas C/aislamiento & purificación , Cisteína/biosíntesis , Cisteína/genética , Cisteína/aislamiento & purificación , Dimerización , Disulfuros/química , Ditiotreitol/química , Humanos , Microscopía Electrónica , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/genética
18.
Diabetes ; 50(7): 1515-21, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11423471

RESUMEN

Mutations in the HNF4alpha gene are responsible for type 1 maturity-onset diabetes of the young (MODY1), which is characterized by a defect in insulin secretion. Hepatocyte nuclear factor (HNF)-4alpha is a transcription factor that plays a critical role in the transcriptional regulation of genes involved in glucose metabolism in both hepatocytes and pancreatic beta-cells. Recent evidence has implicated AMP-activated protein kinase (AMPK) in the modulation of both insulin secretion by pancreatic beta-cells and the control of glucose-dependent gene expression in both hepatocytes and beta-cells. Therefore, the question could be raised as to whether AMPK plays a role in these processes by modulating HNF-4alpha function. In this study, we show that activation of AMPK by 5-amino-4-imidazolecarboxamide riboside (AICAR) in hepatocytes greatly diminished HNF-4alpha protein levels and consequently downregulates the expression of HNF-4alpha target genes. Quantitative evaluation of HNF-4alpha target gene expression revealed diminished mRNA levels for HNF-1alpha, GLUT2, L-type pyruvate kinase, aldolase B, apolipoprotein (apo)-B, and apoCIII. Our data clearly demonstrate that the MODY1/HNF-4alpha transcription factor is a novel target of AMPK in hepatocytes. Accordingly, it can be suggested that in pancreatic beta-cells, AMPK also acts by decreasing HNF-4alpha protein level, and therefore insulin secretion. Hence, the possible role of AMPK in the physiopathology of type 2 diabetes should be considered.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Proteínas de Unión al ADN , Diabetes Mellitus Tipo 1/metabolismo , Complejos Multienzimáticos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/farmacología , Animales , Apolipoproteína C-III , Apolipoproteínas B/biosíntesis , Apolipoproteínas B/genética , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Regulación hacia Abajo , Activación Enzimática , Fructosa-Bifosfato Aldolasa/biosíntesis , Fructosa-Bifosfato Aldolasa/genética , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 2 , Factor Nuclear 4 del Hepatocito , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/enzimología , Hígado/enzimología , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Piruvato Quinasa/biosíntesis , Piruvato Quinasa/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología , Factores de Tiempo , Transcripción Genética
19.
J Leukoc Biol ; 69(4): 645-50, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11310852

RESUMEN

Apolipoprotein C-II (apoC-II), which is known to activate lipoprotein lipase (LPL), was identified by ordered differential display (ODD)-polymerase chain reaction (PCR) as a cDNA fragment exhibiting a distinct increase in expression during 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced differentiation of promonocytic U937 cells into monocytes and macrophages. The amount of apoC-II mRNA expression detectable in U937 cells significantly increased and reached a maximum 24-48 h after treatment with 32 nM TPA. apoC-II mRNA was also detected in monocytic THP-1 cells but was not detected in promyelocytic HL-60 cells. In healthy human tissues, the most significant expression of apoC-II mRNA was in the liver. Although apoC-II mRNA expression was markedly up-regulated during the induced differentiation of HL-60 cells into monocytes and macrophages with 32 nM TPA, such expression was not induced during the differentiation of HL-60 cells into granulocytes with 1.25% dimethyl sulfoxide. These results suggest that human apoC-II expression is induced at the transcription level during myelomonocytic differentiation and may confer an important role to macrophages involved in normal lipid metabolism and atherosclerosis.


Asunto(s)
Apolipoproteínas C/biosíntesis , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células HL-60/efectos de los fármacos , Monocitos/metabolismo , Proteínas de Neoplasias/biosíntesis , Células U937/efectos de los fármacos , Células 3T3/efectos de los fármacos , Células 3T3/metabolismo , Animales , Apolipoproteína C-II , Apolipoproteínas C/genética , Carcinoma/patología , Diferenciación Celular , Ciclina A/biosíntesis , Ciclina A/genética , ADN Complementario/genética , Dimetilsulfóxido/farmacología , Activación Enzimática , Perfilación de la Expresión Génica , Granulocitos/citología , Granulocitos/metabolismo , Células HL-60/citología , Células HL-60/metabolismo , Neoplasias Hematológicas/patología , Humanos , Mucosa Intestinal/metabolismo , Células Jurkat/efectos de los fármacos , Células Jurkat/metabolismo , Cinesinas/biosíntesis , Cinesinas/genética , Metabolismo de los Lípidos , Lipoproteína Lipasa/metabolismo , Hígado/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Monocitos/citología , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Técnica de Sustracción , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Células U937/citología , Células U937/metabolismo
20.
Toxicol Appl Pharmacol ; 170(2): 113-23, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11162775

RESUMEN

It has been proposed that the hepatocellular proliferation induced by peroxisome proliferators may occur through an indirect mechanism involving cytokine release as opposed to direct regulation of cell growth genes by PPARalpha. We compared the induction of peroxisome proliferation and cell proliferation in C57Bl/6 mice treated with 100 mg/kg/day WY14,643 in the presence or absence of increasing doses of dexamethasone (DEX), an inhibitor of the release of proinflammatory cytokines. Biochemical markers of peroxisome proliferation, including fatty acyl-CoA oxidase activity, CYP4A content, and liver-to-body-weight ratios were markedly increased in the WY14,643-treated mice. DEX coadministration, up to a maximum dose of 50 mg/kg/day, did not prevent the induction of these parameters. Acyl-CoA oxidase mRNA levels increased 5-fold with WY14,643 treatment and 15-fold with DEX coadministration at 5 mg/kg/day. ApoCIII mRNA levels were decreased by 50% in WY14,643-treated mice. DEX alone at 5 mg/kg/day increased the ApoCIII mRNA 4-fold, but WY14,643 coadministration also inhibited this induction by greater than 50%. In addition, immunohistochemical detection of peroxisomes with anti-PMP-70 antibody demonstrated marked increase in hepatocellular peroxisomes in WY14,643-treated mice regardless of DEX treatment. In contrast, coadministration of DEX at 2 mg/kg/day partially inhibited the hepatocyte proliferation response (measured by BrdU incorporation or Ki-67 immunohistochemical detection). Moreover, DEX at doses of 5 mg/kg/day or higher completely inhibited the induction of cell proliferation and, at these higher doses, reduced the cell proliferation rate to levels below the vehicle-treated control mice. Our studies clearly demonstrate that the hepatocellular proliferation induced by a peroxisome proliferator can be modulated independently of the other pleiotropic effects usually induced by these agents, suggesting an indirect mechanism of hyperplasia.


Asunto(s)
Antiinflamatorios/farmacología , Dexametasona/farmacología , Proliferadores de Peroxisomas/antagonistas & inhibidores , Pirimidinas/antagonistas & inhibidores , Acil-CoA Oxidasa , Animales , Apolipoproteína C-III , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/genética , Peso Corporal/efectos de los fármacos , División Celular/efectos de los fármacos , Interacciones Farmacológicas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hígado/anatomía & histología , Hígado/citología , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidorreductasas/biosíntesis , Oxidorreductasas/genética , Proliferadores de Peroxisomas/toxicidad , Pirimidinas/toxicidad , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...